Gene Expression Profiling of Desmoid Tumors by

1 downloads 0 Views 495KB Size Report
Mar 28, 2015 - concerning gene-expression analysis in desmoid tumors. One of ... gender, age at diagnosis, location (intra-abdominal, abdominal wall, and ...
Published OnlineFirst April 15, 2015; DOI: 10.1158/1078-0432.CCR-14-2910

Clinical Cancer Research

Biology of Human Tumors

Gene Expression Profiling of Desmoid Tumors by cDNA Microarrays and Correlation with Progression-Free Survival  bastien Salas1,2, Celine Brulard3, Philippe Terrier4, Dominique Ranchere-Vince5, Se Agnes Neuville3, Louis Guillou6, Marick Lae7, Agnes Leroux8, Olivier Verola9, Kurtz Jean-Emmanuel10, Sylvie Bonvalot11, Jean-Yves Blay12, Axel Le Cesne13, Alain Aurias3, Jean-Michel Coindre3,14, and Frederic Chibon3,15

Abstract Purpose: Because desmoid tumors exhibit an unpredictable clinical course, translational research is crucial to identify the predictive factors of progression in addition to the clinical parameters. The main issue is to detect patients who are at a higher risk of progression. The aim of this work was to identify molecular markers that can predict progression-free survival (PFS). Experimental Design: Gene-expression screening was conducted on 115 available independent untreated primary desmoid tumors using cDNA microarray. We established a prognostic gene-expression signature composed of 36 genes. To test robustness, we randomly generated 1,000 36-gene signatures and compared their outcome association to our define 36-genes molecular signature and we calculated pos-

itive predictive value (PPV) and negative predictive value (NPV). Results: Multivariate analysis showed that our molecular signature had a significant impact on PFS while no clinical factor had any prognostic value. Among the 1,000 random signatures generated, 56.7% were significant and none was more significant than our 36-gene molecular signature. PPV and NPV were high (75.58% and 81.82%, respectively). Finally, the top two genes downregulated in no-recurrence were FECH and STOML2 and the top gene upregulated in no-recurrence was TRIP6. Conclusions: By analyzing expression profiles, we have identified a gene-expression signature that is able to predict PFS. This tool may be useful for prospective clinical studies. Clin Cancer Res;

Introduction

(1, 2), but because desmoid tumors exhibit an unpredictable clinical course and an indistinguishable morphology, translational research is crucial to identify the predictive factors of progression in addition to the clinical parameters. A significant improvement would be to be able to detect patients who are at a higher risk of progression and those with no risk of progression. Biologically, alterations of the APC (mutation or loss of the entire locus) and CTNNB1 mutation might constitute an initial mutually exclusive alteration (3, 4). Moreover, Salas and colleagues described three recurrent and relevant alterations of chromosomes 8, 20, and 6 by array comparative genomic hybridization (CGH) array. These alterations could be involved in the same pathway or could confer a selective advantage. Patients harboring CTNNB1 mutations, in particular CTNNB1 (45F) mutations, are at risk of recurrence and the wild-type appears to be a good prognostic marker (5, 6). There have been only a few reports concerning gene-expression analysis in desmoid tumors. One of them demonstrated that a gene-expression signature could distinguish desmoid tumors from nodular fasciitis and suggested that selected tyrosine kinases, transcription factors, and members of the Wnt, TGFb, IFN, and TNF signaling pathways could distinguish these two entities (7). Another study has shown that it was identified genes, in particular ADAM12, WISP-1, and SOX 1, which were uniquely overexpressed in 12 cases of aggressive fibromatosis compared with expression in normal skeletal tissues and a variety of normal tissues. The authors concluded that geneexpression patterns may be useful in the classification of subtypes of aggressive fibromatosis (8). Finally, Colombo and colleagues

Desmoid tumors are mesenchymal fibroblastic/myofibroblastic proliferations. The major obstacle in the management of desmoid tumors is their high propensity for local recurrence even after complete surgical removal. Currently, an initial "wait-and-see" policy is explored as a possible standard of care

1 Aix Marseille Univ, CRO2, INSERM U911, Marseille, France. 2APHM, Timone Hospital, Department of Medicine, Division of Adult Oncology, Marseille, France. 3Department of Pathology, INSERM U916, Bergonie Institute, Bordeaux, France. 4Department of Pathology, Gustave Roussy Institute, Villejuif, France. 5Department of Pathology, Leon Berard Center, Lyon, France. 6University Institute of Pathology, Lausanne, Switzerland. 7Department of Pathology, Institut Curie, Paris, France. 8Department of Pathology, AlexisVautrin Center, Nancy, France. 9Department of Pathology, Saint-Louis Hospital, Paris, France. 10 Department of Oncology and Hematology, University Hospital, Strasbourg, France. 11Department of Surgery, Gustave Roussy Institute, Villejuif, France. 12Department of Medicine, Leon Berard Center, Lyon, France. 13Department of Medicine, Gustave Roussy Institute,  galen University Bordeaux, Bordeaux, Villejuif, France. 14Victor Se France. 15Translational Research, Bergonie Institute, Bordeaux, France.

Note: The data used in this article were provided by the French Sarcoma Group database as part of the ConticaBase (www.conticabase.org). bastien Salas, Aix Marseille University, 13005 Corresponding Author: Se Marseille, France. Phone: 33-4-91-38-57-08; Fax: 33-4-91-38-76-58; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-14-2910 2015 American Association for Cancer Research.

4194

21(18); 4194–200. 2015 AACR.

Clin Cancer Res; 21(18) September 15, 2015

Downloaded from clincancerres.aacrjournals.org on October 20, 2015. © 2015 American Association for Cancer Research.

Published OnlineFirst April 15, 2015; DOI: 10.1158/1078-0432.CCR-14-2910

Expression Data from 115 Desmoid Tumors

Translational Relevance The aim of this study was to identify molecular markers that can predict progression-free-survival (PFS) and thus to distinguish those desmoid tumors for which the use of aggressive treatment is justified rather than a "wait-and-see" strategy. This study clearly demonstrates that there is prognostic molecular signature of desmoid tumors that could benefit from different therapeutic strategies. The main question raised is how patients should be managed. Should patients with poor prognostic molecular signature be operated straightaway in a curative intent and/or benefit from early medical treatment? This study is the starting point for prospective studies, the only way to answer these questions and optimize the management of desmoid tumors. Prospective validation of the molecular signature is under way in France through a clinical trial evaluating the "wait-and-see" strategy (ClinicalTrials.gov identifier NCT01801176).

(9) compared the gene-expression profiles of 14 sporadic desmoid tumors to those of five normal tissues acquired from corresponding desmoid tumor patients and six solitary fibrous tumor specimens. The protein products of three of the upregulated desmoid tumor genes, ADAM12, MMP2, and midkine, were found to be commonly expressed in a large cohort of human desmoid tumor samples assembled on a tissue microarray. Overexpression of midkine was significantly correlated with decreased time to primary recurrence. Moreover, midkine was found to enhance the migration and invasion of primary desmoid tumor cell cultures. These studies suggest the utility of midkine as a clinical desmoid tumor molecular prognosticator and a potential therapeutic target. Midkine could be a novel b-catenin transcriptional target (9). However, no relationship was noted between gene-expression profiling and the clinical course of the disease. In this study, molecular markers predictive for progression-free survival (PFS) were investigated using gene-expression screening that was conducted on 115 available independent untreated primary desmoid tumors using cDNA microarray.

Materials and Methods Patients and samples From February 1, 1987 to March 6, 2008, 115 consecutive patients with sporadic aggressive fibromatosis were diagnosed for their first tumoral event in 16 participating cancer centers. Among tumors in the 115 patients, 66 formed the training samples. The diagnosis of desmoid tumors was confirmed in each case by collegial histologic analysis (mesenchymal fibroblastic/myofibroblastic proliferations). The following clinical data were collected: gender, age at diagnosis, location (intra-abdominal, abdominal wall, and extra-abdominal), size of tumor, and follow-up (Table 1). All patients had an initial surgical resection. Histologic evaluation of surgical margins was available in 91 (79%) cases. Forty-seven patients (41%) had R0 resection, 32 (28%) had R1 resection, and 12 (10%) had R2 resection (macroscopic incomplete as R2 resection; microscopic incomplete resection as R1 resection; microscopic complete resection as R0 resection). Radiotherapy generally included photons or electrons with a median

www.aacrjournals.org

dose of 50 Gray. Forty-three patients received radiotherapy. Surgery was followed by radiotherapy in 16 patients. Clinical and histologic data were entered into a centralized computerized database (www.conticabase.org). All samples were obtained after informed consent from patients. Pathology review Histologic slides of all patients entered in this study were reviewed by the pathology subcommittee of the French Sarcoma Group (GSF). This subcommittee included 20 pathologists and a monthly slide review session was performed. For each tumor, one to eight slides were reviewed collegially. Histologic typing was based on the World Health Organization (WHO) histologic typing of soft tissue tumors. Histopathologic diagnosis was confirmed by the search for CTNNB mutations. All training samples had the CTNNB1 mutation. RNA extraction and cDNA array Total RNAs were extracted from frozen tumor samples with TRizol reagent (Life Technologies, Inc.) and purified using the RNeasy Min Elute TM Cleanup Kit (Qiagen) according to the manufacturer's procedures. RNA quality was checked on an Agilent 2100 bioanalyzer (Agilent Technologies). Samples were then analyzed on Human Genome U133 Plus 2.0 array (Affymetrix), according to the manufacturer's procedures. All microarray data were simultaneously normalized using the GCRMA algorithm (Wu J and Gentry (2014) RIwcfJMJ. GRCMA: Background Adjustment Using Sequence Information. R package version 2.40.0). Minimum information about a microarray experiment–compliant data have been deposited at Gene Expression Omnibus under accession number GSE58697. Gene-expression analysis Differential expression was established using the limma R package and P values were adjusted using the Benjamini– Hochberg procedure (10). Analysis of variance was performed by using GeneSpring GX software (Agilent Technologies) one-way ANOVA test and P values were adjusted by using the Benjamini– Table 1. Patient and disease characteristics at baseline Characteristics

Cohort A Cohort B Cohorts A þ B (n ¼ 66) (n ¼ 49) (n ¼ 115) 2.36 (0.03–12.04) 1.43 (0.33–11.07) 1.82 (0.08–11.97)

Median follow-up (years) (IC 95) Age at diagnosis (%) 37 years >37 years Nd Male sex (%) Location (%) Intra-abdominal Abdominal wall Extra-abdominal Size (%) 7 cm 3 Mutations CTNNB1 (%)

27 37 2 23

(41) (56) (3) (35)

25 (50) 24 (50) 20 (41)

52 (45) 61 (53) 2 (2) 43 (37)

4 (6) 13 (20) 49 (74)

8 (16) 9 (18) 32 (66)

12 (10) 22 (18) 71 (62)

35 (53) 17 (26) 14 (21)

20 (41) 22 (45) 7 (14)

55 (48) 39 (34) 21 (18)

48 (72) 0 13 (20) 5 (8) 0 66 (100)

14 (29) 30 (61) 1 (2) 1 (2) 3 (6) 29 (60)

62 (54) 30 (26) 14 (12) 6 (5) 3 (3) 95(82)

Clin Cancer Res; 21(18) September 15, 2015

Downloaded from clincancerres.aacrjournals.org on October 20, 2015. © 2015 American Association for Cancer Research.

4195

Published OnlineFirst April 15, 2015; DOI: 10.1158/1078-0432.CCR-14-2910

Salas et al.

Hochberg procedure. To assign prognosis, we applied the nearest centroid method. Centroids represent a centered mean of expression for the signature genes for each patient outcome (recurrence and no-recurrence) of the training sets. Each sample of the validation sets was allocated to the prognostic class (centroid) with the highest Spearman correlation. Gene Ontology (GO) analysis was performed to establish statistical enrichment in GO terms by using Gorilla software (11). Statistical analysis PFS is defined as time from the date of initial diagnosis to the date of progression or recurrence or last follow-up. Local recurrence-free survival (LRFS) is defined as time from the date of initial diagnosis to the date of recurrence or last follow-up. We chose to study PFS because the validation cohort consisted of patients who underwent surgery that was not only R0 and R1 but also R2. Survival curves were obtained by the Kaplan–Meier method and compared with the log-rank test. All survival analyses were performed by using R software, version 2.14.1 (R development Core Team, Vienna Austria, 2009) and survival package (Therneau T (2013). A Package for Survival Analysis in S. R package version 2.37-4). The Cox proportional hazards model was used to calculate adjusted hazard ratios (HR) and their 95% confidence intervals (95% CI). Variables with a P value less than 0.05 in univariate analyses were tested in the multivariate analysis. Multivariate analysis was performed by using Cox regression with Firth's correction (R by Meinhard Ploner and Fortran by Georg Heinze, 2012; coxphf: Cox regression with Firth's penalized likelihood. R package, version 1.09). The positive predictive value (PPV) is the ratio of the number of patients having neither a recurrence nor progression at the end of follow-up and classified in the group with a good prognostic molecular signature over the total number of patients classified as having a good prognostic signature. The negative predictive value (NPV) is the ratio of the number of patients whose disease recurred or progressed and were classified as having a poor prognostic signature over the total number of patients classified as having a poor prognostic signature. Sensitivity, specificity, and accuracy were also calculated for the molecular signature.

Results Establishing molecular signature In unsupervised analysis, no molecular signature could be identified. To identify molecular markers that can predict PFS of desmoid tumors, we compared expression profiles in supervised analyses of 48 patients without recurrence versus 18 patients with two or three local recurrences. We identified 18 genes upregulated in recurrence samples and 18 genes downregulated in recurrence samples (Table 2). The supervised analysis comparing patients without recurrence with those having one or more recurrences did not identify any prognostic molecular signature. Classification based on biologic process categories from GO (www.geneontology.org) of the 36 genes demonstrated four enriched GO process terms: cellular component organization or biogenesis (GO:0071840), cellular component organization (GO:0016043), single-organism organelle organization (GO:1902589), organelle organization (GO:0006996), which include 16, 16, 10, and 12 genes, respectively (Table 3). The interaction of the lists of genes from the ANOVA analysis comparing 0-versus-2, 0-versus-3, and 2-versus-3 recurrences identified three genes as shown in the Venn diagram (Fig. 1): FECH, STOML2, and TRIP6. TRIP6 was overexpressed in the good outcome group unlike STOML2 and FECH, which were overexpressed in the poor outcome group. Prognostic factors for PFS and molecular signature validation To test whether clinical factors can predict PFS, we performed univariate analysis for age at diagnosis (cutoff, 37 years), tumor site (intra-abdominal versus abdominal wall versus extraabdominal) and tumor size (cutoff, 7 cm) in the whole cohort. Tumor site and tumor size were not significant but age at diagnosis (P ¼ 4.1e1002) had a significant impact on PFS. Survival analysis was also established for a 36-gene molecular signature with centroid defined on cohort A (n ¼ 66) and classification and validation defined on the whole cohort (n ¼ 115). Univariate analysis showed that the molecular signature predicted PFS (P ¼ 1.7e1007; Fig. 2). The molecular signature also has a prognostic value in LRFS (only the recurrence is considered as an event) in the validation cohort

Table 2. Final gene set containing 36 genes: 18 genes were upregulated in no-recurrence samples and 18 genes were downregulated in no-recurrence samples Probe set id 209129_at 226728_at 204986_s_at 229377_at 206846_s_at 220128_s_at 231767_at 236229_at 203204_s_at 214251_s_at 215692_s_at 236123_at 228929_at 244614_at 232463_at 237317_at 232331_at 234106_s_at

Downregulated genes in recurrence Corrected P value 8.93e05 1.17e04 1.77e04 2.55e04 4.27e04 5.22e04 6.35e04 8.01e04 8.22e04 9.82e04 9.93e04 1.01e03 1.05e03 1.48e03 1.62e03 1.68e03 2.26e03 2.43e03

4196 Clin Cancer Res; 21(18) September 15, 2015

Gene symbol TRIP6 SLC27A1 TAOK2 GRTP1 HDAC6 NPAL2 HOXB4 Hs.661286 JMJD2A NUMA1 MPPED2 ST7L DNASE1 TFG CXYorf10 Hs.127312.0 Hs.25717.0 FLYWCH1

Probe set id 203116_s_at 215416_s_at 225439_at 200014_s_at 226312_at 230465_at 202854_at 211727_s_at 227211_at 229253_at 200006_at 226776_at 203312_x_at 203606_at 200749_at 217880_at 202121_s_at 226596_x_at

Upregulated genes in recurrence Corrected P value 3.23e07 1.09e05 2.49e05 1.34e05 2.26e05 2.73e05 3.58e04 3.66e04 3.70e04 7.60e04 8.07e04 8.22e04 8.27e04 1.40e03 1.47e03 2.69e03 3.17e03 3.32e03

Gene symbol FECH STOML2 NUDCD1 HNRNPC RICTOR HS2ST1 HPRT1 COX11 PHF19 THEM4 PARK7 ENY2 ARF6 NDUFS6 RAN CDC27 CHMP2A LOC729852

Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 20, 2015. © 2015 American Association for Cancer Research.

Published OnlineFirst April 15, 2015; DOI: 10.1158/1078-0432.CCR-14-2910

Expression Data from 115 Desmoid Tumors

Table 3. Classification based on biologic process categories from GO of the 36 genes GO ID Corrected P Z score Number of genes List of genes RAN HPRT1 TRIP6 PHF19 THEM4 ENY2 KDM4A GO:0071840 6.06e103 3.08 16 NUMA1 CHMP2A TAOK2 STOML2 COX11 NDUFS6 RICTOR PARK7 HDAC6 RAN HPRT1 TRIP6 PHF19 THEM4 ENY2 KDM4A GO:0016043 7.24e103 3.10 16 NUMA1 CHMP2A TAOK2 STOML2 COX11 NDUFS6 RICTOR PARK7 HDAC6 NUMA1 RAN STOML2 TAOK2 GO:1902589 8.97e103 6.11 10 NDUFS6 RICTOR THEM4 ENY2 KDM4A HDAC6 NUMA1 RAN STOML2 TAOK2 NDUFS6 4.70 12 PHF19 GO:0006996 1.62e102 RICTOR THEM4 PARK7 ENY2 KDM4A HDAC6

(P ¼ 1.e105). The prognostic value of the three-gene molecular signature (FECH, STOML2, and TRIP6) was lower than that of the 36-gene molecular signature (respectively P ¼ 0.025 and P ¼ 1.7e1007). Multivariate analysis comparing age at diagnosis to the 36-gene molecular signature demonstrated that the latter (P ¼ 7.9e1007) was more predictive than age at diagnosis, which was not significant (Table 4). The molecular signature had also prognostic value in mutated cases whatever the type of mutation (P ¼ 4  108), in S45 cases (P ¼ 3  103

www.aacrjournals.org

), T41 cases (P ¼ 1.3  104), and S45 and T41 cases (P ¼ 2.5  107) but molecular signature has no prognostic value in nonmutated cases (P ¼ 0.59). 36-gene signatures versus random signatures To test its robustness, we randomly generated 1,000 36-gene signatures and compared their outcome-association to our defined 36-gene molecular signature. Exactly the same procedure was used with centroid definition on cohort A and validation in the whole cohort. Among the 1,000 random signatures generated, 56.7% were significant (P < 0.05) and none was more significant (P < 1.7e107) than our 36-gene molecular signature. Finally, the following values were obtained for our 36-gene molecular signature: PPV, 75.58%; NPV, 81.82%; specificity, 72.58%; sensitivity, 81.82%; and accuracy, 78.26%. Prognostic value of the signature in myxofibrosarcomas The signature was also tested in 42 myxofibrosarcomas. These cases were previously published by Chibon and colleagues (12). In this cohort, we found a prognostic value in metastasis-free survival but not in LRFS (respectively P ¼ 0.007 and P ¼ 0.47).

Discussion One of the main problems in managing desmoid tumors is their locoregional aggressiveness and their propensity to recur after initial treatment. The aim of this study was to identify molecular markers that can predict PFS and thus to distinguish those desmoid tumors for which the use of aggressive treatment is justified rather than a "wait-and-see" strategy. A few studies to date have tried to correlate biologic prognostic markers with outcome, but the clinical impact was limited as a result of low robustness (5, 6). Here, we performed the largest gene-expression analysis of sporadic desmoid tumors from the French Sarcoma Group tumor bank ever published. By analyzing expression profiles, we have identified a gene-expression signature that is able to predict PFS. This molecular signature identified two groups with clearly distinct PFS in the two sets of subjects. Patients in the good prognostic group had achieved a 2-year PFS rate of 86% while those in the poor prognostic group had a 2-year PFS rate of 44%. The top two genes upregulated in the recurrence group were FECH and STOML2 and the top gene upregulated in the norecurrence was TRIP6. These genes corresponded to the interaction of the lists of genes obtained from ANOVA analysis comparing 0-versus-2, 0-versus-3, and 2-versus-3 recurrences. The function of these genes of interest is detailed below. The last step of the heme synthetic pathway, which is essential for the cellular energy metabolism, is the incorporation of iron into protoporphyrin IX (PpIX) by the FECH enzyme. 5-Aminolevulinic acid (ALA) is a small-molecule precursor for the synthesis of (PpIX; ref. 13). Loss of FECH enzyme activity might be responsible for enhanced PpIX accumulation in human carcinomas (14). Van Hillegersberg and colleagues (15) have shown that oral administration of ALA resulted in progressive accumulation of protoporphyrin in a rat colon carcinoma but not in the surrounding liver tissue. This team and others suggested that PpIX formation in human cancers, especially as a response to ALA medication, might be related to lowered FECH enzyme activity. Moreover, Kemmner and colleagues (16) were able to demonstrate a significant downregulation of FECH mRNA

Clin Cancer Res; 21(18) September 15, 2015

Downloaded from clincancerres.aacrjournals.org on October 20, 2015. © 2015 American Association for Cancer Research.

4197

Published OnlineFirst April 15, 2015; DOI: 10.1158/1078-0432.CCR-14-2910

Salas et al.

Entity list 3 : One-way ANOVA custom selection 2 vs. 3 3,797 entities

Entity list 1 : One-way ANOVA custom selection 0 vs. 2 80 entities

19 25

5

Figure 1. Venn diagram: interaction of the lists of genes from ANOVA analysis comparing 0-versus-2, 0-versus-3, and 2-versus-3 recurrences.

3 3,770

33

1,179 Entity list 2 : One-way ANOVA custom selection 0 vs. 3 4,985 entities

4198 Clin Cancer Res; 21(18) September 15, 2015

0.8 0.6 0.4 0.2

Progression-free survival

1.0

Ewing sarcoma cells. Although short-term proliferation was not considerably affected by TRIP6 knockdown, silencing of the protein significantly reduced migration, invasion, long-term proliferation, and clonogenicity of Ewing sarcoma cells in vitro as well as tumorigenicity in vivo (30). Moreover, in nasopharyngeal cancer cells (NPC), TRIP6 overexpression/knockdown results in significant enhancement/inhibition of NPC cell migration, respectively (31). Finally, knockdown of TRIP6 in glioblastoma or ovarian cancer xenografts restores nuclear p27 (KIP1) expression and impairs tumor proliferation and may have a significant impact on enhanced NF-kB activity, resistance to apoptosis, and Fas-mediated cell invasion in glioblastomas (29, 32). Paradoxically, in desmoid tumors, TRIP 6 was overexpressed in the good outcome group. However, unlike STOML2 and FECH, TRIP6 is connected to b-catenin in the literature, the main signaling pathway in desmoid tumors. Indeed, TRIP6 might compete with b-catenin for binding with the MAGI-1b/PTEN signalosome to destabilize E-cadherin junctional complexes and to promote cell motility through the regulation of Akt/NF-kB targets and/or effectors of focal adhesion (33). STOML2 and FECH are likely b-catenin transcriptional targets.

P log rank = 1.7e–07

0.0

expression in gastric, colonic, and rectal carcinomas. These data were used as a rationale for the use of photodynamic therapy (PDT) supplemented with exogenously added ALA in particular on urothelial carcinoma (17). In our study, FECH was overexpressed in more aggressive forms of desmoid tumors. Recent evidence indicates that stomatin-like protein 2 (STOML2) regulates mitochondrial functions (i.e., energy production, calcium buffering, and apoptosis) by organizing mitochondrial membranes into defined cardiolipin-enriched microdomains, which then facilitate the optimal assembly of membrane-associated molecular complexes (18). STOML2 has been identified as an oncogenic-related protein and found to be upregulated in multi-cancers. First, STOML2 might serve as a prognostic marker especially in human gallbladder cancer, gastric cancer, glioma, colorectal cancer, breast cancer, and pulmonary squamous cell carcinoma (19–24). STOML2 is also involved in invasion, regulating cell growth and cell adhesion in human esophageal squamous cell carcinoma, glioma, and human endometrial adenocarcinoma (21, 25–27). In desmoid tumor that behaves aggressively by infiltrating tissues deeply, STOML2 overexpression is clinically related to PFS. Thyroid hormone receptor-interacting protein 6 (TRIP6) is a zyxin-related adaptor protein and focal adhesion molecule (28). TRIP6 associates with a variety of molecules from the cell surface to the nucleus, regulates actin reorganization, focal adhesion assembly/disassembly, cell migration/invasion, antiapoptotic signaling, and transcriptional control. Notably, TRIP6 binds to lysophophatidic acid (LPA) receptor 2 (LPA2) and the Fas/CD95 receptor to promote LPA- and Fas ligandinduced cell migration in a c-Src-dependent manner. TRIP6 can also regulate prosurvival signaling via activation of NF-kB, extracellular signal–regulated kinase (ERK), and phosphatidylinositol 3-kinase (PI3K)/AKT, and nuclear TRIP6 acts as a transcriptional coregulator of AP-1 and NF-kB. These data suggest that TRIP6 functions at a point of convergence of multiple signaling pathways critical for cancer development (29). Thus, TRIP6 acts as an oncogene that partially accounts for the autonomous migratory, invasive, and proliferative properties of Ewing sarcoma cells independent of EWS/FLI1. TRIP6 knockdown demonstrated that TRIP6 expression confers a proproliferative and proinvasive transcriptional signature to

0

5

10

15

20

Years Figure 2. Effect of molecular signature on PFS. Patients in good prognostic group had achieved a 2-year PFS rate of 86% while patients in poor prognostic group had a rate of 44%.

Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 20, 2015. © 2015 American Association for Cancer Research.

Published OnlineFirst April 15, 2015; DOI: 10.1158/1078-0432.CCR-14-2910

Expression Data from 115 Desmoid Tumors

Table 4. Univariate and multivariate analysis for prognostic factors in PFS Univariate P HR (95% CI) Molecular signature 1.7e1007 5.3 (2.64–10.58) Age at diagnosis 4.1e1002 1.7 (1.02–3.08)

Classification based on biologic process categories from GO of the 36 genes principally demonstrated four clusters composed of 16, 16, 10, and 12 genes, respectively. These clusters relate to general biologic processes. Validation of the molecular signature analysis was obtained from the whole population including a part of the training. Even though the validation cohort was not totally independent, the prognostic value of this molecular signature was highly significant in this rare disease. In multivariate analysis, only the 36-gene molecular signature was prognostic of PFS, unlike any classical clinical prognostic factor, such as tumor size, tumor site, or age. Moreover, PPVs and NPVs were high, suggesting that this molecular test could be used at the time of diagnosis to stratify patients enrolled in randomized trials. Finally, to test its robustness, we compared its prognostic value to that of randomly generated signatures according to the methodology published in the article by Venet and colleagues. By comparing 47 published breast cancer outcome signatures to signatures of identical size constructed from random genes, they showed that 60% of the published signatures were not significantly better outcome predictors than random signatures (34). In soft tissue sarcoma, the CINSARC signature is particularly robust with only two random signatures more significantly associated with outcomes (35). In this study, our signature was always more significant than random signatures, thus demonstrating the high reliability of our results. Moreover, the fact that the molecular signature had prognostic value in another fibroblastic entity such as myxofibrosarcoma reinforces our idea that this signature is robust and that this signature probably reflects a biologic properties. Although considered nonmalignant owing to their inability to metastasize, the locoregional recurrence rate of desmoid tumors after resection is nearly 50% and the likelihood of accelerating the evolution of the disease with surgery is still debated. This shows that there is an unpredictable clinical course in desmoid tumors and justifies a "wait-and-see" policy as initial strategy. It is therefore necessary to identify patients who are at a higher risk of progression and the search for molecular markers may be a way forward (1, 2). This study clearly demonstrates that there is prognostic molecular signature of desmoid tumors that could benefit from different therapeutic strategies. The main question raised is how patients should be managed. Should patients with poor prognostic molecular signature be operated straightaway in a curative intent and/or benefit from early medical treatment? This study is the starting point for prospective studies, the only way to answer these questions and optimize the management of desmoid tumors.

Multivariate P 7.9e1007 5.1e1001

HR (95% CI) 4.8 (2.50–10.24) 1.2 (0.69–2.14)

Prospective validation of the molecular signature is under way in France through a clinical trial evaluating the "wait-and-see" strategy (ClinicalTrials.gov identifier NCT01801176). However, our work concerns surgical patients and not those in a "wait-andsee" strategy. Thus, we cannot affirm that this molecular signature who predicts progression after surgery, will predict progression before surgery. Finally, somatic markers are not the only factors to be considered because desmoid tumors may completely shrink spontaneously and undergo a change in size over time according to external events (pregnancy, trauma, surgery performed at another site, etc.). The host environment is doubtlessly involved.

Disclosure of Potential Conflicts of Interest No potential conflicts of interest were disclosed.

Authors' Contributions Conception and design: S. Salas, J.-Y. Blay, J.-M. Coindre, F. Chibon Development of methodology: S. Salas, A. Aurias, J.-M. Coindre, F. Chibon Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): S. Salas, P. Terrier, D. Ranchere-Vince, A. Neuville, L. Guillou, M. Lae, A. Leroux, O. Verola, K. Jean-Emmanuel, S. Bonvalot, J.-Y. Blay, A. Le Cesne, J.-M. Coindre, F. Chibon Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S. Salas, C. Brulard, L. Guillou, J.-Y. Blay, A. Le Cesne, F. Chibon Writing, review, and/or revision of the manuscript: S. Salas, C. Brulard, A. Neuville, M. Lae, A. Leroux, S. Bonvalot, J.-Y. Blay, J.-M. Coindre, F. Chibon Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): S. Salas, P. Terrier, L. Guillou, J.-Y. Blay, J.-M. Coindre, F. Chibon Study supervision: S. Salas, A. Aurias, J.-M. Coindre, F. Chibon

Acknowledgments The authors thank R. Cooke for editing the English. They also thank the patient advocacy group, SOS desmoids, and NetSARC, RREPS, LYRIC, DevWECAN, and EUROSARC networks for their support.

Grant Support This work has been financially supported by La Ligue National Contre le Cancer. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. Received December 16, 2014; revised March 19, 2015; accepted March 28, 2015; published OnlineFirst April 15, 2015.

References 1. Gronchi A, Colombo C, Le Pechoux C, Dei Tos AP, Le Cesne A, Marrari A, et al. Sporadic desmoid-type fibromatosis: a stepwise approach to a non-metastasising neoplasm–a position paper from the Italian and the French Sarcoma Group. Ann Oncol 2014;25: 578–83.

www.aacrjournals.org

2. Bonvalot S, Desai A, Coppola S, Le Pechoux C, Terrier P, Domont J, et al. The treatment of desmoid tumors: a stepwise clinical approach. Ann Oncol 2012;23(Suppl 10):x158–66. 3. Salas S, Chibon F, Noguchi T, Terrier P, Ranchere-Vince D, Lagarde P, et al. Molecular characterization by array comparative genomic hybridization

Clin Cancer Res; 21(18) September 15, 2015

Downloaded from clincancerres.aacrjournals.org on October 20, 2015. © 2015 American Association for Cancer Research.

4199

Published OnlineFirst April 15, 2015; DOI: 10.1158/1078-0432.CCR-14-2910

Salas et al.

4.

5.

6.

7.

8. 9.

10.

11.

12.

13. 14.

15.

16.

17.

18.

and DNA sequencing of 194 desmoid tumors. Genes Chromosomes Cancer 2010;49:560–8. Nieuwenhuis MH, Casparie M, Mathus-Vliegen LM, Dekkers OM, Hogendoorn PC, Vasen HF. A nation-wide study comparing sporadic and familial adenomatous polyposis-related desmoid-type fibromatoses. Int J Cancer 2011;129:256–61. Lazar AJ, Tuvin D, Hajibashi S, Habeeb S, Bolshakov S, MayordomoAranda E, et al. Specific mutations in the beta-catenin gene (CTNNB1) correlate with local recurrence in sporadic desmoid tumors. Am J Pathol 2008;173:1518–27. Domont J, Salas S, Lacroix L, Brouste V, Saulnier P, Terrier P, et al. High frequency of beta-catenin heterozygous mutations in extra-abdominal fibromatosis: a potential molecular tool for disease management. Br J Cancer 2010;102:1032–6. Bacac M, Migliavacca E, Stehle JC, McKee T, Delorenzi M, Coindre JM, et al. A gene expression signature that distinguishes desmoid tumours from nodular fasciitis. J Pathol 2006;208:543–53. Skubitz KM, Skubitz AP. Gene expression in aggressive fibromatosis. J Lab Clin Med 2004;143:89–98. Colombo C, Creighton CJ, Ghadimi MP, Bolshakov S, Warneke CL, Zhang Y, et al. Increased midkine expression correlates with desmoid tumour recurrence: a potential biomarker and therapeutic target. J Pathol 2011;225:574–82. Smyth GK, Michaud J, Scott HS. Use of within-array replicate spots for assessing differential expression in microarray experiments. Bioinformatics 2005;21:2067–75. Eden E, Navon R, Steinfeld I, Lipson D, Yakhini Z. GOrilla: a tool for discovery and visualization of enriched GO terms in ranked gene lists. BMC Bioinformatics 2009;10:48. Chibon F, Lagarde P, Salas S, Perot G, Brouste V, Tirode F, et al. Validated prediction of clinical outcome in sarcomas and multiple types of cancer on the basis of a gene expression signature related to genome complexity. Nat Med 2010;16:781–7. Rao AU, Carta LK, Lesuisse E, Hamza I. Lack of heme synthesis in a freeliving eukaryote. Proc Natl Acad Sci U S A 2005;102:4270–5. Dailey HA, Smith A. Differential interaction of porphyrins used in photoradiation therapy with ferrochelatase. Biochem J 1984;223: 441–5. Van Hillegersberg R, Van den Berg JW, Kort WJ, Terpstra OT, Wilson JH. Selective accumulation of endogenously produced porphyrins in a liver metastasis model in rats. Gastroenterology 1992;103:647–51. Kemmner W, Wan K, Ruttinger S, Ebert B, Macdonald R, Klamm U, et al. Silencing of human ferrochelatase causes abundant protoporphyrin-IX accumulation in colon cancer. FASEB J 2008;22:500–9. Inoue K, Fukuhara H, Kurabayashi A, Furihata M, Tsuda M, Nagakawa K, et al. Photodynamic therapy involves an antiangiogenic mechanism and is enhanced by ferrochelatase inhibitor in urothelial carcinoma. Cancer Sci 2013;104:765–72. Christie DA, Kirchhof MG, Vardhana S, Dustin ML, Madrenas J. Mitochondrial and plasma membrane pools of stomatin-like protein 2 coalesce at the immunological synapse during T cell activation. PLoS ONE 2012;7: e37144.

4200 Clin Cancer Res; 21(18) September 15, 2015

19. Wang WX, Lin QF, Shen D, Liu SP, Mao WD, Ma G, et al. Clinicopathological significance of SLP-2 overexpression in human gallbladder cancer. Tumour Biol 2014;35:419–23. 20. Li XH, He F, Yan SM, Li Y, Cao Y, Huang CY, et al. Increased expression of stomatin-like protein 2 (STOML2) predicts decreased survival in gastric adenocarcinoma: a retrospective study. Med Oncol 2014;31:763. 21. Song L, Liu L, Wu Z, Lin C, Dai T, Yu C, et al. Knockdown of stomatinlike protein 2 (STOML2) reduces the invasive ability of glioma cells through inhibition of the NF-kappaB/MMP-9 pathway. J Pathol 2012;226:534–43. 22. Yeoh LC, Loh CK, Gooi BH, Singh M, Gam LH. Hydrophobic protein in colorectal cancer in relation to tumor stages and grades. World J Gastroenterol 2010;16:2754–63. 23. Chang D, Ma K, Gong M, Cui Y, Liu ZH, Zhou XG, et al. SLP-2 overexpression is associated with tumour distant metastasis and poor prognosis in pulmonary squamous cell carcinoma. Biomarkers 2010;15:104–10. 24. Cao W, Zhang B, Liu Y, Li H, Zhang S, Fu L, et al. High-level SLP-2 expression and HER-2/neu protein expression are associated with decreased breast cancer patient survival. Am J Clin Pathol 2007;128:430–6. 25. Cao W, Zhang B, Ding F, Zhang W, Sun B, Liu Z. Expression of SLP-2 was associated with invasion of esophageal squamous cell carcinoma. PLoS ONE 2013;8:e63890. 26. Zhang L, Ding F, Cao W, Liu Z, Liu W, Yu Z, et al. Stomatin-like protein 2 is overexpressed in cancer and involved in regulating cell growth and cell adhesion in human esophageal squamous cell carcinoma. Clin Cancer Res 2006;12:1639–46. 27. Cui Z, Zhang L, Hua Z, Cao W, Feng W, Liu Z. Stomatin-like protein 2 is overexpressed and related to cell growth in human endometrial adenocarcinoma. Oncol Rep 2007;17:829–33. 28. Lin VT, Lin FT. TRIP6: an adaptor protein that regulates cell motility, antiapoptotic signaling and transcriptional activity. Cell Signal 2011; 23:1691–7. 29. Lin VT, Lin VY, Lai YJ, Chen CS, Liu K, Lin WC, et al. TRIP6 regulates p27 KIP1 to promote tumorigenesis. Mol Cell Biol 2013;33:1394–409. 30. Grunewald TG, Willier S, Janik D, Unland R, Reiss C, Prazeres da Costa O, et al. The Zyxin-related protein thyroid receptor interacting protein 6 (TRIP6) is overexpressed in Ewing's sarcoma and promotes migration, invasion and cell growth. Biol Cell 2013;105:535–47. 31. Fei J, Li J, Shen S, Zhou W. Characterization of TRIP6-dependent nasopharyngeal cancer cell migration. Tumour Biol 2013;34:2329–35. 32. Lai YJ, Lin VT, Zheng Y, Benveniste EN, Lin FT. The adaptor protein TRIP6 antagonizes Fas-induced apoptosis but promotes its effect on cell migration. Mol Cell Biol 2010;30:5582–96. 33. Chastre E, Abdessamad M, Kruglov A, Bruyneel E, Bracke M, Di Gioia Y, et al. TRIP6, a novel molecular partner of the MAGI-1 scaffolding molecule, promotes invasiveness. FASEB J 2009;23:916–28. 34. Venet D, Dumont JE, Detours V. Most random gene expression signatures are significantly associated with breast cancer outcome. PLoS Comput Biol 2011;7:e1002240. 35. Brulard C, Chibon F. Robust gene expression signature is not merely a significant P value. Eur J Cancer 2013;49:2771–3.

Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 20, 2015. © 2015 American Association for Cancer Research.

Published OnlineFirst April 15, 2015; DOI: 10.1158/1078-0432.CCR-14-2910

Gene Expression Profiling of Desmoid Tumors by cDNA Microarrays and Correlation with Progression-Free Survival Sébastien Salas, Celine Brulard, Philippe Terrier, et al. Clin Cancer Res 2015;21:4194-4200. Published OnlineFirst April 15, 2015.

Updated version

Cited articles

E-mail alerts Reprints and Subscriptions Permissions

Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-14-2910

This article cites 35 articles, 11 of which you can access for free at: http://clincancerres.aacrjournals.org/content/21/18/4194.full.html#ref-list-1

Sign up to receive free email-alerts related to this article or journal. To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at [email protected]. To request permission to re-use all or part of this article, contact the AACR Publications Department at [email protected].

Downloaded from clincancerres.aacrjournals.org on October 20, 2015. © 2015 American Association for Cancer Research.